Cargando…

Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19

Metabolic pathways drive cellular behavior. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes lung tissue damage directly by targeting cells or indirectly by producing inflammatory cytokines. However, whether functional alterations are related to metabolic changes in lung...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Shuangyan, Zhao, Fuxiaonan, Ye, Jing, Li, Kuan, Wang, Qi, Du, Zhongchao, Yue, Qing, Wang, Sisi, Wu, Qi, Chen, Huaiyong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Berlin Heidelberg 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8755516/
https://www.ncbi.nlm.nih.gov/pubmed/35022857
http://dx.doi.org/10.1007/s00430-021-00727-0
_version_ 1784632394652844032
author Li, Shuangyan
Zhao, Fuxiaonan
Ye, Jing
Li, Kuan
Wang, Qi
Du, Zhongchao
Yue, Qing
Wang, Sisi
Wu, Qi
Chen, Huaiyong
author_facet Li, Shuangyan
Zhao, Fuxiaonan
Ye, Jing
Li, Kuan
Wang, Qi
Du, Zhongchao
Yue, Qing
Wang, Sisi
Wu, Qi
Chen, Huaiyong
author_sort Li, Shuangyan
collection PubMed
description Metabolic pathways drive cellular behavior. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes lung tissue damage directly by targeting cells or indirectly by producing inflammatory cytokines. However, whether functional alterations are related to metabolic changes in lung cells after SARS-CoV-2 infection remains unknown. Here, we analyzed the lung single-nucleus RNA-sequencing (snRNA-seq) data of several deceased COVID-19 patients and focused on changes in transcripts associated with cellular metabolism. We observed upregulated glycolysis and oxidative phosphorylation in alveolar type 2 progenitor cells, which may block alveolar epithelial differentiation and surfactant secretion. Elevated inositol phosphate metabolism in airway progenitor cells may promote neutrophil infiltration and damage the lung barrier. Further, multiple metabolic alterations in the airway goblet cells are associated with impaired muco-ciliary clearance. Increased glycolysis, oxidative phosphorylation, and inositol phosphate metabolism not only enhance macrophage activation but also contribute to SARS-CoV-2 induced lung injury. The cytotoxicity of natural killer cells and CD8(+) T cells may be enhanced by glycerolipid and inositol phosphate metabolism. Glycolytic activation in fibroblasts is related to myofibroblast differentiation and fibrogenesis. Glycolysis, oxidative phosphorylation, and glutathione metabolism may also boost the aging, apoptosis and proliferation of vascular smooth muscle cells, resulting in pulmonary arterial hypertension. In conclusion, this preliminary study revealed a possible cellular metabolic basis for the altered innate immunity, adaptive immunity, and niche cell function in the lung after SARS-CoV-2 infection. Therefore, patients with COVID-19 may benefit from therapeutic strategies targeting cellular metabolism in future. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00430-021-00727-0.
format Online
Article
Text
id pubmed-8755516
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Springer Berlin Heidelberg
record_format MEDLINE/PubMed
spelling pubmed-87555162022-01-13 Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19 Li, Shuangyan Zhao, Fuxiaonan Ye, Jing Li, Kuan Wang, Qi Du, Zhongchao Yue, Qing Wang, Sisi Wu, Qi Chen, Huaiyong Med Microbiol Immunol Original Investigation Metabolic pathways drive cellular behavior. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes lung tissue damage directly by targeting cells or indirectly by producing inflammatory cytokines. However, whether functional alterations are related to metabolic changes in lung cells after SARS-CoV-2 infection remains unknown. Here, we analyzed the lung single-nucleus RNA-sequencing (snRNA-seq) data of several deceased COVID-19 patients and focused on changes in transcripts associated with cellular metabolism. We observed upregulated glycolysis and oxidative phosphorylation in alveolar type 2 progenitor cells, which may block alveolar epithelial differentiation and surfactant secretion. Elevated inositol phosphate metabolism in airway progenitor cells may promote neutrophil infiltration and damage the lung barrier. Further, multiple metabolic alterations in the airway goblet cells are associated with impaired muco-ciliary clearance. Increased glycolysis, oxidative phosphorylation, and inositol phosphate metabolism not only enhance macrophage activation but also contribute to SARS-CoV-2 induced lung injury. The cytotoxicity of natural killer cells and CD8(+) T cells may be enhanced by glycerolipid and inositol phosphate metabolism. Glycolytic activation in fibroblasts is related to myofibroblast differentiation and fibrogenesis. Glycolysis, oxidative phosphorylation, and glutathione metabolism may also boost the aging, apoptosis and proliferation of vascular smooth muscle cells, resulting in pulmonary arterial hypertension. In conclusion, this preliminary study revealed a possible cellular metabolic basis for the altered innate immunity, adaptive immunity, and niche cell function in the lung after SARS-CoV-2 infection. Therefore, patients with COVID-19 may benefit from therapeutic strategies targeting cellular metabolism in future. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00430-021-00727-0. Springer Berlin Heidelberg 2022-01-13 2022 /pmc/articles/PMC8755516/ /pubmed/35022857 http://dx.doi.org/10.1007/s00430-021-00727-0 Text en © The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2022 This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.
spellingShingle Original Investigation
Li, Shuangyan
Zhao, Fuxiaonan
Ye, Jing
Li, Kuan
Wang, Qi
Du, Zhongchao
Yue, Qing
Wang, Sisi
Wu, Qi
Chen, Huaiyong
Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title_full Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title_fullStr Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title_full_unstemmed Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title_short Cellular metabolic basis of altered immunity in the lungs of patients with COVID-19
title_sort cellular metabolic basis of altered immunity in the lungs of patients with covid-19
topic Original Investigation
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8755516/
https://www.ncbi.nlm.nih.gov/pubmed/35022857
http://dx.doi.org/10.1007/s00430-021-00727-0
work_keys_str_mv AT lishuangyan cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT zhaofuxiaonan cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT yejing cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT likuan cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT wangqi cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT duzhongchao cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT yueqing cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT wangsisi cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT wuqi cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19
AT chenhuaiyong cellularmetabolicbasisofalteredimmunityinthelungsofpatientswithcovid19